Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Juri Na is active.

Publication


Featured researches published by Juri Na.


Molecular Cancer Therapeutics | 2015

Targeting BRG1 Chromatin Remodeler via Its Bromodomain for Enhanced Tumor Cell Radiosensitivity In Vitro and In Vivo

Su-Jung Kwon; Seul-Ki Lee; Juri Na; Shin-Ai Lee; Han-Sae Lee; Ji-Hye Park; June-Key Chung; Hyewon Youn; Jongbum Kwon

Radiotherapy treats cancer by inducing DNA double-strand breaks (DSB) in tumor cells using ionizing radiation. However, DNA repair in tumor cells often leads to radioresistance and unsuccessful outcome. Inhibition of DNA repair by targeting repair proteins can increase radiosensitivity of tumor cells. The BRG1 chromatin remodeling enzyme assists DSB repair by stimulating γ-H2AX formation and BRG1 binding to acetylated histones at DSBs via bromodomain (BRD) is critical for this activity. Here, we show that ectopic expression of BRG1-BRD inhibited γ-H2AX and DSB repair after irradiation and increased the radiosensitivity in various human cancer cells, including HT29 colon cancer. Dimerization of BRG1-BRD, increasing its chromatin binding affinity, aggravated the defects in γ-H2AX and DSB repair and further enhanced the radiosensitivity. While little affecting the upstream ATM activation, BRG1-BRD in irradiated HT29 cells inhibited the recruitment of 53BP1 to damaged chromatin, the downstream event of γ-H2AX, and compromised the G2–M checkpoint and increased apoptosis. Importantly, in a xenograft mouse model, BRG1-BRD increased the radiosensitivity of HT29 tumors, which was further enhanced by dimerization. These data suggest that BRG1-BRD radiosensitizes tumor cells by a dominant negative activity against BRG1, which disrupts γ-H2AX and its downstream 53BP1 pathways, leading to inefficient DNA repair, G2–M checkpoint defect, and increased apoptosis. This work therefore identifies BRG1-BRD as a novel tumor radiosensitizer and its action mechanism, providing the first example of chromatin remodeler as a target for improving cancer radiotherapy. Mol Cancer Ther; 14(2); 597–607. ©2014 AACR.


Journal of Immunology | 2016

GM-CSF Induces Inflammatory Macrophages by Regulating Glycolysis and Lipid Metabolism

Yi Rang Na; Gyo Jung Gu; Daun Jung; Young Won Kim; Juri Na; Jin Sun Woo; Joo Youn Cho; Hyewon Youn; Seung-Hyeok Seok

GM-CSF induces proinflammatory macrophages, but the underlying mechanisms have not been studied thus far. In this study, we investigated the mechanisms of how GM-CSF induces inflammatory macrophages. First, we observed that GM-CSF increased the extent of LPS-induced acute glycolysis in murine bone marrow–derived macrophages. This directly correlates with an inflammatory phenotype because glycolysis inhibition by 2-deoxyglucose abolished GM-CSF–mediated increase of TNF-α, IL-1β, IL-6, and IL-12p70 synthesis upon LPS stimulation. Increased glycolytic capacity is due to de novo synthesis of glucose transporter (GLUT)-1, -3, and -4, as well as c-myc. Meanwhile, GM-CSF increased 3-hydroxy-3-methyl-glutaryl-CoA reductase, which is the rate-limiting enzyme of the mevalonate pathway. Inhibition of acute glycolysis or 3-hydroxy-3-methyl-glutaryl-CoA reductase abrogated the inflammatory effects of GM-CSF priming in macrophages. Finally, mice with inflamed colons exposed to dextran sodium sulfate containing GLUT-1high macrophages led to massive uptake of [18F]-fluorodeoxyglucose, but GM-CSF neutralization reduced the positron-emission tomography signal in the intestine and also decreased GLUT-1 expression in colonic macrophages. Collectively, our results reveal glycolysis and lipid metabolism created by GM-CSF as the underlying metabolic constructs for the function of inflammatory macrophages.


Hepatology | 2015

Interaction of tetraspan(in) TM4SF5 with CD44 promotes self‐renewal and circulating capacities of hepatocarcinoma cells

Doohyung Lee; Juri Na; Jihye Ryu; Hye Jin Kim; Seo Hee Nam; Minkyung Kang; Jae Woo Jung; Mi‑Sook Lee; Haeng Eun Song; Jungeun Choi; Gyu Ho Lee; T.Y. Kim; June-Key Chung; Ki Hun Park; Sung Hak Kim; Hyunggee Kim; Howon Seo; Pilhan Kim; Hyewon Youn; Jung Weon Lee

Tumor metastasis involves circulating and tumor‐initiating capacities of metastatic cancer cells. Epithelial‐mesenchymal transition (EMT) is related to self‐renewal capacity and circulating tumor cell (CTC) characteristics for tumor metastasis. Although tumor metastasis is a life‐threatening, complicated process that occurs through circulation of tumor cells, mechanistic aspects of self‐renewal and circulating capacities have been largely unknown. Hepatic transmembrane 4 L six family member 5 (TM4SF5) promotes EMT for malignant growth and migration, so it was rationalized that TM4SF5, as a hepatocellular carcinoma (HCC) biomarker, might be important for metastatic potential. Here, self‐renewal capacity by TM4SF5 was mechanistically explored using hepatocarcinoma cells with or without TM4SF5 expression, and we explored whether they became CTCs using mouse liver‐orthotopic model systems. We found that TM4SF5‐dependent sphere growth correlated with CD24−, aldehyde dehydrogenase (ALDH) activity, as well as a physical association between CD44 and TM4SF5. Interaction between TM4SF5 and CD44 was through their extracellular domains with N‐glycosylation modifications. TM4SF5/CD44 interaction activated proto‐oncogene tyrosine‐protein kinase Src (c‐Src)/signal transducer and activator of transcription 3 (STAT3)/Twist‐related protein 1 (Twist1)/B‐cell‐specific Moloney murine leukemia virus integration site 1 (Bmi1) signaling for spheroid formation, whereas disturbing the interaction, expression, or activity of any component in this signaling pathway inhibited spheroid formation. In serial xenografts using 200∼5,000 cells per injection, TM4SF5‐positive tumors exhibited subpopulations with locally increased CD44 expressions, supporting for tumor cell differentiation. TM4SF5‐positive, but not TM4SF5‐ or CD44‐knocked‐down, cells were identified circulating in blood 4‐6 weeks after orthotopic liver injection using in vivo laser scanning endomicroscopy. Anti‐TM4SF5 reagent blocked their metastasis to distal intestinal organs. Conclusion: TM4SF5 promotes self‐renewal and CTC properties supported by TM4SF5+/CD44+(TM4SF5‐bound)/ALDH+/CD24− markers during HCC metastasis. (Hepatology 2015;61:1978‐1997)


Theranostics | 2016

Dihydropyrimidine Dehydrogenase Is a Prognostic Marker for Mesenchymal Stem Cell-Mediated Cytosine Deaminase Gene and 5-Fluorocytosine Prodrug Therapy for the Treatment of Recurrent Gliomas

Taemoon Chung; Juri Na; Young-Il Kim; Da-Young Chang; Young Il Kim; Hyeonjin Kim; Ho Eun Moon; Keon Wook Kang; Dong Soo Lee; June-Key Chung; Sung-Soo Kim; Haeyoung Suh-Kim; Sun Ha Paek; Hyewon Youn

We investigated a therapeutic strategy for recurrent malignant gliomas using mesenchymal stem cells (MSC), expressing cytosine deaminase (CD), and prodrug 5-Fluorocytosine (5-FC) as a more specific and less toxic option. MSCs are emerging as a novel cell therapeutic agent with a cancer-targeting property, and CD is considered a promising enzyme in cancer gene therapy which can convert non-toxic 5-FC to toxic 5-Fluorouracil (5-FU). Therefore, use of prodrug 5-FC can minimize normal cell toxicity. Analyses of microarrays revealed that targeting DNA damage and its repair is a selectable option for gliomas after the standard chemo/radio-therapy. 5-FU is the most frequently used anti-cancer drug, which induces DNA breaks. Because dihydropyrimidine dehydrogenase (DPD) was reported to be involved in 5-FU metabolism to block DNA damage, we compared the survival rate with 5-FU treatment and the level of DPD expression in 15 different glioma cell lines. DPD-deficient cells showed higher sensitivity to 5-FU, and the regulation of DPD level by either siRNA or overexpression was directly related to the 5-FU sensitivity. For MSC/CD with 5-FC therapy, DPD-deficient cells such as U87MG, GBM28, and GBM37 showed higher sensitivity compared to DPD-high U373 cells. Effective inhibition of tumor growth was also observed in an orthotopic mouse model using DPD- deficient U87MG, indicating that DPD gene expression is indeed closely related to the efficacy of MSC/CD-mediated 5-FC therapy. Our results suggested that DPD can be used as a biomarker for selecting glioma patients who may possibly benefit from this therapy.


Theranostics | 2015

Codon-optimized Human Sodium Iodide Symporter (opt-hNIS) as a Sensitive Reporter and Efficient Therapeutic Gene

Young-Hwa Kim; Hyewon Youn; Juri Na; Kee-Jong Hong; Keon Wook Kang; Dong Soo Lee; June-Key Chung

To generate a more efficient in vivo reporter and therapeutic gene, we optimized the coding sequence of the human sodium/iodide symporter (NIS) gene by replacing NIS DNA codons from wild type to new codons having the highest usage in human gene translation. The Codon Adaptation Index (CAI), representing the number of codons effective for human expression, was much improved (0.79 for hNIS, 0.97 for opt-hNIS). Both wild-type (hNIS) and optimized human NIS (opt-hNIS) were cloned into pcDNA3.1 and pMSCV vectors for transfection. Various cancer cell lines such as thyroid (TPC-1, FRO, B-CPAP), breast (MDA-MB-231), liver (Hep3B), cervical (HeLa), and glioma (U87MG) were transfected with pcDNA3.1/hNIS or pcDNA3.1/opt-hNIS. 125I uptake by opt-hNIS-expressing cells was 1.6 ~ 2.1 times higher than uptake by wild-type hNIS-expressing cells. Stable cell lines were also established by retroviral transduction using pMSCV/hNIS or pMSCV/opt-hNIS, revealing higher NIS protein levels and 125I uptake in opt-hNIS-expressing cells than in hNIS-expressing cells. Moreover, scintigraphic images from cell plates and mouse xenografts showed stronger signals from opt-hNIS-expressing cells than hNIS-expressing cells, and radioactivity uptake by opt-hNIS-expressing tumors was 2.3-fold greater than that by hNIS-expressing tumors. To test the efficacy of radioiodine therapy, mouse xenograft models were established with cancer cells expressing hNIS or opt-hNIS. 131I treatment reduced tumor sizes of hNIS- and opt-hNIS-expressing tumors to 0.57- and 0.27- fold, respectively, compared to their sizes before therapy, suggesting an improved therapeutic effect of opt-hNIS. In summary, this study shows that codon optimization strongly increases hNIS protein levels and radioiodine uptake, thus supporting opt-hNIS as a more sensitive reporter and efficient therapeutic gene.


Journal of Immunology | 2017

Correction: GM-CSF Induces Inflammatory Macrophages by Regulating Glycolysis and Lipid Metabolism

Yi Rang Na; Gyo Jeong Gu; Daun Jung; Young Won Kim; Juri Na; Jin Sun Woo; Joo Youn Cho; Hyewon Youn; Seung-Hyeok Seok

Na, Y. R., G. J. Gu, D. Jung, Y. W. Kim, J. Na, J. S. Woo, J. Y. Cho, H. Youn, and S. H. Seok. 2016. GM-CSF induces inflammatory macrophages by regulating glycolysis and lipid metabolism. J . Immunol . 197: [4101–4109][1]. The second author’s middle name was published incorrectly. The correct


Molecular Imaging | 2015

Relationship between Apoptosis Imaging and Radioiodine Therapy in Tumor Cells with Different Sodium Iodide Symporter Gene Expression

Kyung Oh Jung; Hyewon Youn; Young-Hwa Kim; Seunghoo Kim; Juri Na; Yong-Il Kim; Jin Woo Park; Keon Wook Kang; Dong Soo Lee; June-Key Chung

The therapeutic efficacy of radioiodine (¹³¹I) therapy has been reported to be variable among cancer patients and even between metastatic regions in the same patients. Because the expression level of sodium iodide symporter (NIS) cannot reflect the efficacy of therapy, other strategies are required to predict the precise therapeutic effect of ¹³¹I therapy. In this research, we investigated the correlation between iodine (I) uptake, apoptosis imaging, and therapeutic efficacy. Two HT29 cell lines, cytomegalovirus (CMV)-NIS (or NIS+++) and TERT-NIS (or NIS+), were established by retroviral transfection. I uptake was estimated by I-uptake assay and gamma camera imaging. Apoptosis was evaluated by confocal microscopy and a Maestro fluorescence imaging system (CRi Inc., Woburn, MA) using ApoFlamma (BioACTs, Seoul, Korea), a fluorescent dye-conjugated apoptosis-targeting peptide 1 (ApoPep-1). Therapeutic efficacy was determined by tumor size. The CMV-NIS showed higher I uptake and ApoFlamma signals than TERT-NIS. In xenograft models, CMV-NIS also showed high 99m technetium signals and ApoFlamma signals. Tumor reduction had a stronger correlation with apoptosis imaging signals than with gamma camera imaging signals, which reflect I uptake. Higher NIS-expressing tumors showed increased apoptosis and I uptake, resulting in a significant tumor reduction. Moreover, tumor reduction showed a strong correlation with ApoFlamma imaging compared to I-uptake imaging.The therapeutic efficacy of radioiodine (131I) therapy has been reported to be variable among cancer patients and even between metastatic regions in the same patients. Because the expression level of sodium iodide symporter (NIS) cannot reflect the efficacy of therapy, other strategies are required to predict the precise therapeutic effect of 131I therapy. In this research, we investigated the correlation between iodine (I) uptake, apoptosis imaging, and therapeutic efficacy. Two HT29 cell lines, cytomegalovirus (CMV)-NIS (or NIS+++) and TERT-NIS (or NIS+), were established by retroviral transfection. I uptake was estimated by I-uptake assay and gamma camera imaging. Apoptosis was evaluated by confocal microscopy and a Maestro fluorescence imaging system (CRi Inc., Woburn, MA) using ApoFlamma (BioACTs, Seoul, Korea), a fluorescent dye–conjugated apoptosis-targeting peptide 1 (ApoPep-1). Therapeutic efficacy was determined by tumor size. The CMV-NIS showed higher I uptake and ApoFlamma signals than TERT-NIS. In xenograft models, CMV-NIS also showed high 99m technetium signals and ApoFlamma signals. Tumor reduction had a stronger correlation with apoptosis imaging signals than with gamma camera imaging signals, which reflect I uptake. Higher NIS-expressing tumors showed increased apoptosis and I uptake, resulting in a significant tumor reduction. Moreover, tumor reduction showed a strong correlation with ApoFlamma imaging compared to I-uptake imaging.


International Journal of Endocrinology | 2017

Thyroid-Related Protein Expression in the Human Thymus

Mi Jeong Kim; So Won Oh; Hyewon Youn; Juri Na; Keon Wook Kang; Do Joon Park; Young Joo Park; Ja June Jang; Kyu Eun Lee; Kyeong Cheon Jung; June-Key Chung

Radioiodine whole body scan (WBS), related to sodium iodide symporter (NIS) function, is widely used to detect recurrence/metastasis in postoperative patients with thyroid cancer. However, the normal thymic uptake of radioiodine has occasionally been observed in young patients. We evaluated the expression of thyroid-related genes and proteins in the human thymus. Thymic tissues were obtained from 22 patients with thyroid cancer patients of all ages. The expression of NIS, thyroid-stimulating hormone receptor (TSHR), thyroperoxidase (TPO), and thyroglobulin (Tg) was investigated using immunohistochemistry and quantitative RT-PCR. NIS and TSHR were expressed in 18 (81.8%) and 19 samples (86.4%), respectively, whereas TPO was expressed in five samples (22.7%). Three thyroid-related proteins were localized to Hassalls corpuscles and thymocytes. In contrast, Tg was detected in a single patient (4.5%) localized to vascular endothelial cells. The expression of thyroid-related proteins was not increased in young thymic tissues compared to that in old thymic tissues. In conclusion, the expression of NIS and TSHR was detected in the majority of normal thymus samples, whereas that of TPO was detected less frequently, and that of Tg was detected rarely. The increased thymic uptake of radioiodine in young patients is not due to the increased expression of NIS.


The Journal of Nuclear Medicine | 2016

Radiosensitization effect of bromodomain overexpression on thyroid anaplastic tumor using human applicable low iodine-131 dose

Juri Na; Hyewon Youn; Keon Wook Kang; June-Key Chung


The Journal of Nuclear Medicine | 2016

Evaluating anti-inflammatory effect of alkaline protease using 18F-FDG PET in mouse inflammatory bowel disease

Juri Na; Young Hwa Kim; Michael J. Parmely; Kee-Jong Hong; Keon Wook Kang; Hyewon Youn; June-Key Chung

Collaboration


Dive into the Juri Na's collaboration.

Top Co-Authors

Avatar

Hyewon Youn

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

June-Key Chung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Keon Wook Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Dong Soo Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Mi Jeong Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Daun Jung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jin Sun Woo

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joo Youn Cho

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Kee-Jong Hong

International Vaccine Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge